Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Clin Genet ; 105(4): 453-454, 2024 04.
Article in English | MEDLINE | ID: mdl-38072398

ABSTRACT

This graphic abstract combines pedigree, dysmorphology features, radiographs, and the PRKG2 protein domain, specifically the CNB-A regulatory domain, which harbors a mutation resulting in premature protein termination.


Subject(s)
Exome , Family , Humans , Cyclic GMP-Dependent Protein Kinase Type II/genetics , Exome/genetics , Mutation/genetics , Pedigree
2.
BMC Genom Data ; 24(1): 2, 2023 01 04.
Article in English | MEDLINE | ID: mdl-36600198

ABSTRACT

BACKGROUND: Previous studies have shown that the protein kinase cGMP-dependent 2 (PRKG2) gene is associated with dwarfism in humans, dogo Argentines, and Angus cattle, as well as with height and osteoblastogenesis in humans. Therefore, the PRKG2 gene was used as the target gene to explore whether this gene is associated with several thoracolumbar vertebrae and carcass traits in Dezhou donkeys. RESULTS: In this study, fifteen SNPs were identified by targeted sequencing, all of which were located in introns of the PRKG2 gene. Association analysis illustrated that the g.162153251 G > A, g.162156524 C > T, g.162158453 C > T and, g.162163775 T > G were significantly different from carcass weight. g.162166224 G > A, g.162166654 T > A, g.162167165 C > A, g.162167314 A > C and, g.162172653 G > C were significantly associated with the number of thoracic vertebrae. g.162140112 A > G was significantly associated with the number and the length of lumbar vertebrae, and g.162163775 T > G was significantly associated with the total number of thoracolumbar vertebrae. CONCLUSION: Overall, the results of this study suggest that PRKG2 gene polymorphism can be used as a molecular marker to breed high-quality Dezhou donkeys.


Subject(s)
Cyclic GMP-Dependent Protein Kinase Type II , Equidae , Polymorphism, Single Nucleotide , Animals , Cyclic GMP-Dependent Protein Kinase Type II/genetics , Equidae/genetics , Introns , Phenotype , Polymorphism, Single Nucleotide/genetics , Spine
3.
Carcinogenesis ; 43(6): 584-593, 2022 06 27.
Article in English | MEDLINE | ID: mdl-35188962

ABSTRACT

A large body of evidence has demonstrated that cyclic-guanosine monophosphate (cGMP), signaling has anti-tumor effects that might be used for colon cancer prevention. The tumor-suppressive mechanism and the signaling components downstream of cGMP remain largely unknown. The present study has characterized the expression of cGMP-dependent protein kinases (PKG1, PKG2) in normal and cancerous tissue from human colon. PKG1 was detected in both normal and tumor tissue, where it localized exclusively to the lamina propria and stroma (respectively). In contrast, PKG2 localized specifically to the epithelium where its expression decreased markedly in tumors compared to matched normal tissue. Neither PKG isoform was detected at the RNA or protein level in established colon cancer cell lines. To test for a potential tumor-suppressor role of PKG2 in the colon epithelium, Prkg2 knockout (KO) mice were subjected to azoxymethane/dextran sulfate-sodium (AOM/DSS) treatment. PKG2 deficiency was associated with crypt hyperplasia (Ki67) and almost twice the number of polyps per mouse as wild-type (WT) siblings. In vitro culture of mouse colon epithelium as organoids confirmed that PKG2 was the only isoform expressed, and it was detected in both proliferating and differentiating epithelial compartments. Colon organoids derived from Prkg2 KO mice proliferated more rapidly and exhibited a reduced ability to differentiate compared to WT controls. Taken together our results highlight PKG2 as the central target of cGMP in the colon, where it suppresses carcinogenesis by controlling proliferation in an epithelial-cell intrinsic manner.


Subject(s)
Colon , Colonic Neoplasms , Cyclic GMP-Dependent Protein Kinase Type II , Animals , Azoxymethane , Carcinogenesis/pathology , Cell Proliferation , Colon/pathology , Colonic Neoplasms/pathology , Cyclic GMP/metabolism , Cyclic GMP-Dependent Protein Kinase Type II/genetics , Dextran Sulfate , Epithelium/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout
4.
J Med Genet ; 59(1): 28-38, 2022 01.
Article in English | MEDLINE | ID: mdl-33106379

ABSTRACT

BACKGROUND: C-type natriuretic peptide (CNP), its endogenous receptor, natriuretic peptide receptor-B (NPR-B), as well as its downstream mediator, cyclic guanosine monophosphate (cGMP) dependent protein kinase II (cGKII), have been shown to play a pivotal role in chondrogenic differentiation and endochondral bone growth. In humans, biallelic variants in NPR2, encoding NPR-B, cause acromesomelic dysplasia, type Maroteaux, while heterozygous variants in NPR2 (natriuretic peptide receptor 2) and NPPC (natriuretic peptide precursor C), encoding CNP, cause milder phenotypes. In contrast, no variants in cGKII, encoded by the protein kinase cGMP-dependent type II gene (PRKG2), have been reported in humans to date, although its role in longitudinal growth has been clearly demonstrated in several animal models. METHODS: Exome sequencing was performed in two girls with severe short stature due to acromesomelic limb shortening, brachydactyly, mild to moderate platyspondyly and progressively increasing metaphyseal alterations of the long bones. Functional characterisation was undertaken for the identified variants. RESULTS: Two homozygous PRKG2 variants, a nonsense and a frameshift, were identified. The mutant transcripts are exposed to nonsense-mediated decay and the truncated mutant cGKII proteins, partially or completely lacking the kinase domain, alter the downstream mitogen activation protein kinase signalling pathway by failing to phosphorylate c-Raf 1 at Ser43 and subsequently reduce ERK1/2 activation in response to fibroblast growth factor 2. They also downregulate COL10A1 and upregulate COL2A1 expression through SOX9. CONCLUSION: In conclusion, we have clinically and molecularly characterised a new acromesomelic dysplasia, acromesomelic dysplasia, PRKG2 type (AMDP).


Subject(s)
Cyclic GMP-Dependent Protein Kinase Type II/genetics , Dwarfism/genetics , Mutation , Osteochondrodysplasias/genetics , Brachydactyly , Child , Dwarfism/metabolism , Female , Humans , Osteochondrodysplasias/metabolism , Pedigree , Exome Sequencing
6.
Respir Res ; 22(1): 308, 2021 Dec 04.
Article in English | MEDLINE | ID: mdl-34863181

ABSTRACT

BACKGROUND: Tracheal injury is a common clinical condition that still lacks an effective therapy at present. Stimulation of epithelial sodium channel (ENaC) increases Na+ transport, which is a driving force to keep tracheal mucosa free edema fluid during tracheal injury. Ferulic acid (FA) has been proved to be effective in many respiratory diseases through exerting anti-oxidant, anti-inflammatory, and anti-thrombotic effects. However, these studies rarely involve the level of ion transport, especially ENaC. METHODS: C57BL/J male mice were treated intraperitoneally with normal saline or FA (100 mg/kg) 12 h before, and 12 h after intratracheal administration of lipopolysaccharide (LPS, 5 mg/kg), respectively. The effects of FA on tracheal injury were not only assessed through HE staining, immunofluorescence assay, and protein/mRNA expressions of ENaC located on tracheas, but also evaluated by the function of ENaC in mouse tracheal epithelial cells (MTECs). Besides, to explore the detailed mechanism about FA involved in LPS-induced tracheal injury, the content of cyclic guanosine monophosphate (cGMP) was measured, and Rp-cGMP (cGMP inhibitor) or cGMP-dependent protein kinase II (PKGII)-siRNA (siPKGII) were applied in primary MTECs, respectively. RESULTS: Histological examination results demonstrated that tracheal injury was obviously attenuated by pretreatment of FA. Meanwhile, FA could reverse LPS-induced reduction of both protein/mRNA expressions and ENaC activity. ELISA assay verified cGMP content was increased by FA, and administration of Rp-cGMP or transfection of siPKGII could reverse the FA up-regulated ENaC protein expression in MTECs. CONCLUSIONS: Ferulic acid can attenuate LPS-induced tracheal injury through up-regulation of ENaC at least partially via the cGMP/PKGII pathway, which may provide a promising new direction for preventive and therapeutic strategy in tracheal injury.


Subject(s)
Acute Lung Injury/genetics , Coumaric Acids/pharmacology , Cyclic GMP-Dependent Protein Kinase Type II/genetics , Epithelial Sodium Channels/genetics , Gene Expression Regulation , Trachea/drug effects , Acute Lung Injury/chemically induced , Acute Lung Injury/drug therapy , Animals , Cells, Cultured , Cyclic GMP-Dependent Protein Kinase Type II/biosynthesis , Enzyme-Linked Immunosorbent Assay , Epithelial Sodium Channels/biosynthesis , Free Radical Scavengers/pharmacology , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred C57BL , RNA/genetics , Signal Transduction , Trachea/metabolism , Trachea/pathology
7.
Genes (Basel) ; 12(10)2021 09 24.
Article in English | MEDLINE | ID: mdl-34680883

ABSTRACT

Dwarfism phenotypes occur in many species and may be caused by genetic or environmental factors. In this study, we investigated a family of nine Dogo Argentino dogs, in which two dogs were affected by disproportionate dwarfism. Radiographs of an affected dog revealed a decreased level of endochondral ossification in its growth plates, and a premature closure of the distal ulnar physes. The pedigree of the dogs presented evidence of monogenic autosomal recessive inheritance; combined linkage and homozygosity mapping assigned the most likely position of a potential genetic defect to 34 genome segments, totaling 125 Mb. The genome of an affected dog was sequenced and compared to 795 control genomes. The prioritization of private variants revealed a clear top candidate variant for the observed dwarfism. This variant, PRKG2:XM_022413533.1:c.1634+1G>T, affects the splice donor site and is therefore predicted to disrupt the function of the PKRG2 gene encoding protein, kinase cGMP-dependent type 2, a known regulator of chondrocyte differentiation. The genotypes of the PRKG2 variant were perfectly associated with the phenotype in the studied family of dogs. PRKG2 loss-of-function variants were previously reported to cause disproportionate dwarfism in humans, cattle, mice, and rats. Together with the comparative data from other species, our data strongly suggest PRKG2:c.1634+1G>T to be a candidate causative variant for the observed dwarfism phenotype in Dogo Argentino dogs.


Subject(s)
Cyclic GMP-Dependent Protein Kinase Type II/genetics , Dog Diseases/genetics , Dwarfism/genetics , Genetic Predisposition to Disease , Animals , Cattle , Dog Diseases/pathology , Dogs , Dwarfism/pathology , Dwarfism/veterinary , Genetic Linkage/genetics , Genotype , Humans , Mice , Mutation/genetics , Pedigree , Phenotype , Protein Isoforms/genetics , Rats
8.
Int J Mol Sci ; 22(1)2020 Dec 23.
Article in English | MEDLINE | ID: mdl-33374621

ABSTRACT

Mutations in the CNGA3 gene, which encodes the A subunit of the cyclic guanosine monophosphate (cGMP)-gated cation channel in cone photoreceptor outer segments, cause total colour blindness, also referred to as achromatopsia. Cones lacking this channel protein are non-functional, accumulate high levels of the second messenger cGMP and degenerate over time after induction of ER stress. The cell death mechanisms that lead to loss of affected cones are only partially understood. Here, we explored the disease mechanisms in the Cnga3 knockout (KO) mouse model of achromatopsia. We found that another important effector of cGMP, the cGMP-dependent protein kinase 2 (Prkg2) is crucially involved in cGMP cytotoxicity of cones in Cnga3 KO mice. Virus-mediated knockdown or genetic ablation of Prkg2 in Cnga3 KO mice counteracted degeneration and preserved the number of cones. Analysis of markers of endoplasmic reticulum stress and unfolded protein response confirmed that induction of these processes in Cnga3 KO cones also depends on Prkg2. In conclusion, we identified Prkg2 as a novel key mediator of cone photoreceptor degeneration in achromatopsia. Our data suggest that this cGMP mediator could be a novel pharmacological target for future neuroprotective therapies.


Subject(s)
Color Vision Defects/etiology , Color Vision Defects/metabolism , Cyclic GMP-Dependent Protein Kinase Type II/metabolism , Cyclic Nucleotide-Gated Cation Channels/deficiency , Retinal Cone Photoreceptor Cells/metabolism , Animals , Biomarkers , Color Vision Defects/pathology , Cyclic GMP/metabolism , Cyclic GMP-Dependent Protein Kinase Type II/genetics , Disease Models, Animal , Disease Susceptibility , Endoplasmic Reticulum Stress , Fluorescent Antibody Technique , Gene Expression , Mice , Mice, Knockout , Microscopy, Confocal , Models, Biological , Retinal Degeneration/etiology , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Unfolded Protein Response
9.
J BUON ; 25(2): 729-735, 2020.
Article in English | MEDLINE | ID: mdl-32521860

ABSTRACT

PURPOSE: This research tried to explore the expression level of II cGMP-dependent protein kinase (PKG2) in human ovarian tissue and to clarify the molecular mechanism of EGFR regulation and its clinical significance. METHODS: The expression levels of PKG2 and EGFR in 10 normal ovarian tissues, 14 benign ovarian tumor tissues and 39 epithelial ovarian cancer tissues preserved in the archives of the Affiliated Hospital of Xuzhou Medical University from 2016 to 2018 were detected by real-time fluorescence quantitative (RT-PCR), and the correlation between the expressions of the two genes was analyzed. The expressions of in vitro cultured ovarian cancer cell lines SKOV3, PKG2 and EGFR were detected by RT-PCR and western blot, and the over-expressed PKG2 plasmid and PKG2 small interfering RNA (siRNA) were transfected into the cells, and the protein and phosphorylation of Akt and ERK in EGFR and its downstream signaling pathway were detected by western blot. RESULTS: Compared with normal ovarian tissue, the mRNA and protein expression levels of PKG2 in ovarian cancer tissue and SKOV3 cell line were significantly reduced (p<0.05). However, the mRNA and protein expression levels of EGFR in ovarian cancer tissue and SKOV3 cell line were both high (p<0.05). In addition, after transient transfection of PKG2, the expression changes of PKG2 significantly affected the expression of EGFR, and PKG2 over-expression could significantly inhibit the phosphorylation of Akt and ERK in EGFR and its downstream signaling pathways, thereby affecting cell proliferation. CONCLUSION: PKG2 may play a role in inhibiting EGFR expression in ovarian cancer, but the specific mechanism of its effect on tumor development still needs to be further explored.


Subject(s)
Carcinoma, Ovarian Epithelial/metabolism , Cyclic GMP-Dependent Protein Kinase Type II/metabolism , Ovarian Neoplasms/metabolism , Carcinoma, Ovarian Epithelial/enzymology , Carcinoma, Ovarian Epithelial/genetics , Cell Line, Tumor , Cyclic GMP-Dependent Protein Kinase Type II/biosynthesis , Cyclic GMP-Dependent Protein Kinase Type II/genetics , ErbB Receptors/genetics , ErbB Receptors/metabolism , Female , Humans , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Transfection
10.
Biochim Biophys Acta Mol Cell Res ; 1867(8): 118732, 2020 08.
Article in English | MEDLINE | ID: mdl-32360667

ABSTRACT

Nitric oxide is an important neuromodulator in the CNS, and its production within neurons is modulated by NMDA receptors and requires a fine-tuned availability of L-arginine. We have previously shown that globally inhibiting protein synthesis mobilizes intracellular L-arginine "pools" in retinal neurons, which concomitantly enhances neuronal nitric oxide synthase-mediated nitric oxide production. Activation of NMDA receptors also induces local inhibition of protein synthesis and L-arginine intracellular accumulation through calcium influx and stimulation of eucariotic elongation factor type 2 kinase. We hypothesized that protein synthesis inhibition might also increase intracellular L-arginine availability to induce nitric oxide-dependent activation of downstream signaling pathways. Here we show that nitric oxide produced by inhibiting protein synthesis (using cycloheximide or anisomycin) is readily coupled to AKT activation in a soluble guanylyl cyclase and cGKII-dependent manner. Knockdown of cGKII prevents cycloheximide or anisomycin-induced AKT activation and its nuclear accumulation. Moreover, in retinas from cGKII knockout mice, cycloheximide was unable to enhance AKT phosphorylation. Indeed, cycloheximide also produces an increase of ERK phosphorylation which is abrogated by a nitric oxide synthase inhibitor. In summary, we show that inhibition of protein synthesis is a previously unanticipated driving force for nitric oxide generation and activation of downstream signaling pathways including AKT and ERK in cultured retinal cells. These results may be important for the regulation of synaptic signaling and neuronal development by NMDA receptors as well as for solving conflicting data observed when using protein synthesis inhibitors for studying neuronal survival during development as well in behavior and memory studies.


Subject(s)
Cyclic GMP-Dependent Protein Kinase Type II/metabolism , Nitric Oxide/metabolism , Protein Synthesis Inhibitors/pharmacology , Retina/metabolism , Signal Transduction/drug effects , Animals , Arginine/metabolism , Cell Survival/drug effects , Cells, Cultured , Chick Embryo , Chickens , Cyclic GMP-Dependent Protein Kinase Type II/genetics , Elongation Factor 2 Kinase/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , Nitrates/metabolism , Nitric Oxide Synthase Type I/metabolism , Nitrites , Phosphorylation
11.
Am J Physiol Cell Physiol ; 317(6): C1115-C1127, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31509448

ABSTRACT

In the early phase of pregnancy, decidualization is an indispensable event after mammal embryo implantation, accompanied by proliferation and differentiation of uterine stromal cells. Type II cGMP-dependent protein kinase (Prkg2) belongs to the family of serine/threonine kinase, which plays multiple roles in cellular signaling pathways to control proliferation and differentiation. However, the regulatory function and molecular mechanism of Prkg2 in decidualization are still unknown. In this study, we show that Prkg2 has a gradually increased expression pattern during peri-implantation and artificial decidualization, and the expression of Prkg2 is induced by estrogen and progesterone in the ovariectomized mouse uteri and primary cultured uterine stromal cells, the process of which is blocked by treating with estrogen receptor (ER) antagonist (ICI-182,780) and progesterone receptor (PR) antagonist (RU-486). Inhibition of Prkg2 activity by HA-100 promotes uterine stromal cell proliferation but compromises decidualization with decreased expression of prolactin family 8, subfamily a, member 2. In addition, the functional regulation of decidualization by Prkg2 is accomplished by its induced phosphorylation of glycogen synthase kinase-3ß (GSK-3ß) at serine-9, which results in accumulation of ß-catenin in the decidual cells. Taken together, our findings demonstrate that estrogen and progesterone upregulate the expression of Prkg2 in uterine stromal cells depending on ER and PR; Prkg2 promotes phosphorylation of GSK-3ß at serine-9 and inactivates it, leading to the accumulation of ß-catenin and promoting the process of decidualization. In addition to revealing the regulatory mechanism of Prkg2 that ensures the success of uterine decidualization, our findings will contribute to the understanding in the maintenance of early pregnancy.


Subject(s)
Cyclic AMP/metabolism , Cyclic GMP-Dependent Protein Kinase Type II/genetics , Decidua/metabolism , Stromal Cells/metabolism , beta Catenin/genetics , Animals , Cell Proliferation/drug effects , Cyclic GMP-Dependent Protein Kinase Type II/antagonists & inhibitors , Cyclic GMP-Dependent Protein Kinase Type II/metabolism , Decidua/cytology , Decidua/drug effects , Estrogens/pharmacology , Female , Fulvestrant/pharmacology , Gene Expression Regulation , Glycogen Synthase Kinase 3 beta/genetics , Glycogen Synthase Kinase 3 beta/metabolism , Isoquinolines/pharmacology , Mice , Mifepristone/pharmacology , Ovariectomy , Phosphorylation , Pregnancy , Primary Cell Culture , Progesterone/pharmacology , Prolactin/analogs & derivatives , Prolactin/genetics , Prolactin/metabolism , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Receptors, Progesterone/antagonists & inhibitors , Receptors, Progesterone/genetics , Receptors, Progesterone/metabolism , Signal Transduction , Stromal Cells/cytology , Stromal Cells/drug effects , Sulfonamides/pharmacology , Uterus/cytology , Uterus/drug effects , Uterus/metabolism , beta Catenin/metabolism
12.
Forensic Sci Int Genet ; 36: 50-59, 2018 09.
Article in English | MEDLINE | ID: mdl-29933125

ABSTRACT

DNA methylation is the most extensively studied epigenetic signature, with a large number of studies reporting age-correlated CpG sites in overlapping genes. However, most of these studies lack sample coverage of individuals under 18 years old and therefore little is known about the progression of DNA methylation patterns in children and adolescents. In the present study we aimed to select candidate age-correlated DNA methylation markers based on public datasets from Illumina BeadChip arrays and previous publications, then to explore the resulting markers in 209 blood samples from donors aged between 2 to 18 years old using the EpiTYPER® DNA methylation analysis system. Results from our analyses identified six genes highly correlated with age in the young, in particular the gene KCNAB3, which indicates its potential as a highly informative and specific age biomarker for childhood and adolescence. We outline a preliminary age prediction model based on quantile regression that uses data from the six CpG sites most strongly correlated with age ranges extended to include children and adolescents.


Subject(s)
Aging/genetics , DNA Methylation , Forensic Genetics/methods , Genetic Markers , Acetyltransferases/genetics , Adolescent , Amidohydrolases/genetics , Child , Child, Preschool , CpG Islands/genetics , Cyclic GMP-Dependent Protein Kinase Type II/genetics , Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Edar-Associated Death Domain Protein/genetics , Fatty Acid Elongases , Humans , LIM-Homeodomain Proteins/genetics , Muscle Proteins/genetics , Nerve Tissue Proteins/genetics , Polymerase Chain Reaction , Shaker Superfamily of Potassium Channels , Shaw Potassium Channels/genetics , Software , Transcription Factors/genetics
13.
J Endocrinol ; 238(3): 203-219, 2018 09.
Article in English | MEDLINE | ID: mdl-29914933

ABSTRACT

NO/cGMP signaling is important for bone remodeling in response to mechanical and hormonal stimuli, but the downstream mediator(s) regulating skeletal homeostasis are incompletely defined. We generated transgenic mice expressing a partly-activated, mutant cGMP-dependent protein kinase type 2 (PKG2R242Q) under control of the osteoblast-specific Col1a1 promoter to characterize the role of PKG2 in post-natal bone formation. Primary osteoblasts from these mice showed a two- to three-fold increase in basal and total PKG2 activity; they proliferated faster and were resistant to apoptosis compared to cells from WT mice. Male Col1a1-Prkg2R242Q transgenic mice had increased osteoblast numbers, bone formation rates and Wnt/ß-catenin-related gene expression in bone and a higher trabecular bone mass compared to their WT littermates. Streptozotocin-induced type 1 diabetes suppressed bone formation and caused rapid bone loss in WT mice, but male transgenic mice were protected from these effects. Surprisingly, we found no significant difference in bone micro-architecture or Wnt/ß-catenin-related gene expression between female WT and transgenic mice; female mice of both genotypes showed higher systemic and osteoblastic NO/cGMP generation compared to their male counterparts, and a higher level of endogenous PKG2 activity may be responsible for masking effects of the PKG2R242Q transgene in females. Our data support sexual dimorphism in Wnt/ß-catenin signaling and PKG2 regulation of this crucial pathway in bone homeostasis. This work establishes PKG2 as a key regulator of osteoblast proliferation and post-natal bone formation.


Subject(s)
Bone Diseases, Metabolic/genetics , Bone and Bones/pathology , Cyclic GMP-Dependent Protein Kinase Type II/physiology , Osteogenesis/genetics , Animals , Bone Density/genetics , Bone Diseases, Metabolic/metabolism , Bone and Bones/metabolism , Cell Differentiation/genetics , Cell Proliferation/genetics , Cells, Cultured , Cyclic GMP-Dependent Protein Kinase Type II/genetics , Diabetes Complications/genetics , Diabetes Complications/metabolism , Diabetes Complications/pathology , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Organ Size/genetics , Osteoblasts/metabolism , Osteoblasts/physiology
14.
Med Sci Monit ; 24: 1997-2002, 2018 Apr 04.
Article in English | MEDLINE | ID: mdl-29617357

ABSTRACT

BACKGROUND The present work was performed to detect the potential inhibitory effect of cyclic guanosine monophosphate (cGMP)-dependent protein kinase II (PKG II) on epidermal growth factor (EGF) receptor-induced biological activity and related signal cascades in osteosarcoma cells. MATERIAL AND METHODS We transfected the osteosarcoma MG-63 cell line with an adenoviral vector encoding PKG II cDNA (Ad-PKGII) and incubated the transfected cells with 250 µM 8-pCPT-cGMP to activate the PKG II. We stimulated the MG-63 cells with100 ng/ml EGF, and then detected their proliferation using a CCK-8 assay. Transwell assay was used to examine MG-63 cell migration; and Western blot analysis was used to detect expression of matrix metalloproteinase 9 (MMP-9) and activation of ERK and Akt. RESULTS Stimulating cells by 100 ng/ml EGF promoted MG-63 cell proliferation and migration, ERK and Akt phosphorylation, and MMP-9 expression. These effects of EGF were inhibited in MG-63 cells infected with Ad-PKGII and incubated with 8-pCPT-cGMP. CONCLUSIONS Our results demonstrate that Ad-PKGII infection significantly inhibited EGF-induced proliferation and migration, as well as the associated-signal cascades; which indicates that PKG II might be a potential anti-cancer factor.


Subject(s)
Bone Neoplasms/metabolism , Cyclic GMP-Dependent Protein Kinase Type II/metabolism , Epidermal Growth Factor/antagonists & inhibitors , ErbB Receptors/antagonists & inhibitors , Osteosarcoma/metabolism , Bone Neoplasms/enzymology , Bone Neoplasms/pathology , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Cyclic GMP/metabolism , Cyclic GMP-Dependent Protein Kinase Type II/biosynthesis , Cyclic GMP-Dependent Protein Kinase Type II/genetics , Epidermal Growth Factor/metabolism , Epidermal Growth Factor/pharmacology , ErbB Receptors/metabolism , Humans , Osteosarcoma/enzymology , Osteosarcoma/pathology , Phosphorylation , Protein Binding , Signal Transduction , Transfection
15.
Cell Biol Int ; 42(4): 435-442, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29150923

ABSTRACT

Epidermal growth factor receptor (EGFR) plays an important role in gastric cancer (GC) progression. Our previous data demonstrated that type II cGMP-dependent protein kinase (PKG II) could block the EGF-EGFR axis as well as down-stream signaling pathways, for example, MAPK, PI3 K, and PLC in GC cells. However, the exact mechanisms of PKG II against cancer remain unclear. Therefore, the present work was to address the above question. Human GC cell line AGS was infected with adenoviral construct encoding cDNA of PKG II (Ad-PKG II) to up-regulate PKG II and then treated with 8-pCPT-cGMP. Two-dimensional electrophoresis (2-DE) was used to analyze the changes of protein expression in the cells. The results showed that 17 proteins had more than twofold changes in EGF-treated group compared with control. However, Ad-PKG II could effectively reversed the changes. Furthermore, far upstream element-binding protein 1 (FUBP1) and MarvelD3 were chosen and PKG II activation reversed EGF/EGFR-induced up-regulation of FUBP1 and downregulation of MarvelD3, respectively. MarvelD3 silence effectively abolished the inhibitory effect of PKG II on EGF-triggered migration. These data indicated that the inhibitory effect of PKG II partially was associated with MarvelD3.


Subject(s)
Cyclic GMP-Dependent Protein Kinase Type II/administration & dosage , Epidermal Growth Factor/pharmacology , Stomach Neoplasms/metabolism , Cell Line, Tumor , Cyclic GMP/analogs & derivatives , Cyclic GMP/pharmacology , Cyclic GMP-Dependent Protein Kinase Type II/biosynthesis , Cyclic GMP-Dependent Protein Kinase Type II/genetics , Cyclic GMP-Dependent Protein Kinase Type II/metabolism , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Electrophoresis, Gel, Two-Dimensional , Epidermal Growth Factor/metabolism , ErbB Receptors/metabolism , HEK293 Cells , Humans , Membrane Proteins/metabolism , Phosphorylation/drug effects , RNA-Binding Proteins , Signal Transduction/drug effects , Stomach Neoplasms/genetics , Thionucleotides/pharmacology , Transcriptional Activation
16.
Mol Med Rep ; 16(4): 5729-5737, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28849123

ABSTRACT

Previous studies have indicated that type II cyclic guanosine monophosphate (cGMP)­dependent protein kinase (PKG II) could inhibit the proliferation and migration of gastric cancer cells. However, the effects of PKG II on the biological functions of other types of cancer cells remain to be elucidated. Therefore, the aim of the present study was to investigate the effects of PKG II on cancer cells derived from various types of human tissues, including A549 lung, HepG2 hepatic, OS­RC­2 renal, SW480 colon cancer cells and U251 glioma cells. Cancer cells were infected with adenoviral constructs coding PKG II (Ad­PKG II) to up­regulate PKG II expression, and treated with 8­(4­chlorophenylthio) (8­pCPT)­cGMP to activate the kinase. A Cell Counting kit 8 assay was used to detect cell proliferation. Cell migration was measured using a Transwell assay, whereas a terminal deoxynucleotidyl transferase 2'­deoxyuridine, 5'­triphosphate nick­end labeling assay was used to detect cell apoptosis. A pull­down assay was used to investigate the activation of Ras­related C3 botulinum toxin substrate (Rac) 1 and western blotting was used to detect the expression of proteins of interest. The present results demonstrated that EGF (100 ng/ml, 24 h) promoted the proliferation and migration of cancer cells, and it suppressed their apoptosis. In addition, treatment with EGF enhanced the activation of Rac1, and up­regulated the protein expression of proliferating cell nuclear antigen, matrix metalloproteinase (MMP)2, MMP7 and B­cell lymphoma (Bcl)­2, whereas it down­regulated the expression of Bcl­2­associated X protein. Transfection of cancer cells with Ad­PKG II, and PKG II activation with 8­pCPT­cGMP, was identified to counteract the effects triggered by EGF. The present results suggested that PKG II may exert inhibitory effects on the proliferation and migration of various types of cancer cells.


Subject(s)
Cyclic GMP-Dependent Protein Kinase Type II/metabolism , Neoplasms/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/drug effects , Cyclic GMP-Dependent Protein Kinase Type II/genetics , Epidermal Growth Factor/metabolism , Epidermal Growth Factor/pharmacology , Gene Expression , Humans , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 7/metabolism , Neoplasms/genetics , Neoplasms/pathology , Signal Transduction , rac1 GTP-Binding Protein/metabolism
17.
Hum Pathol ; 67: 198-204, 2017 09.
Article in English | MEDLINE | ID: mdl-28412213

ABSTRACT

To the best of our knowledge, this manuscript describes clinical and pathologic findings of the first case of acute mast cell leukemia harboring t(4;5)(q21;q33), compatible with fusion of the PDGFRB gene to a rare partner, PRKG2. Translocation involving the PDGFRB gene is confirmed by fluorescence in situ hybridization study. This case presented a relatively fulminant clinical course with acute mast cell leukemia and "C" findings (cytopenia, hepatosplenomegaly, and weight loss), mast cell sarcoma, and severe basophilia. Despite aggressive presentation initially, the patient responded well to tyrosine kinase inhibitor treatment and is currently in complete remission 33 months after diagnosis. This case significantly extends the disease spectrum associated with PRKG2/PDGFRB fusion gene. Recognizing the whole spectrum of diseases associated with this fusion is critical because tyrosine kinase inhibitor treatment has been exceedingly effective in these patients.


Subject(s)
Biomarkers, Tumor/genetics , Chromosomes, Human, Pair 4 , Chromosomes, Human, Pair 5 , Leukemia, Mast-Cell/genetics , Translocation, Genetic , Acute Disease , Antineoplastic Agents/therapeutic use , Biopsy , Cyclic GMP-Dependent Protein Kinase Type II/genetics , Gene Fusion , Genetic Predisposition to Disease , Humans , Imatinib Mesylate/therapeutic use , Immunohistochemistry , Leukemia, Mast-Cell/drug therapy , Leukemia, Mast-Cell/enzymology , Leukemia, Mast-Cell/pathology , Male , Middle Aged , Molecular Diagnostic Techniques , Molecular Targeted Therapy , Phenotype , Protein Kinase Inhibitors/therapeutic use , Receptor, Platelet-Derived Growth Factor beta/genetics , Remission Induction , Time Factors , Treatment Outcome
18.
Biochem Biophys Res Commun ; 483(1): 82-87, 2017 01 29.
Article in English | MEDLINE | ID: mdl-28057484

ABSTRACT

Although type II cGMP-dependent protein kinase (PKGII) is a major downstream effector of cGMP in chondrocytes and attenuates the FGF receptor 3/ERK signaling pathway, its direct target proteins have not been fully explored. In the present study, we attempted to identify PKGII-targeted proteins, which are associated with the inhibition of FGF-induced MAPK activation. Although FGF2 stimulation induced the phosphorylation of ERK1/2, MEK1/2, and Raf-1 at Ser-338 in rat chondrosarcoma cells, pretreatment with a cell-permeable cGMP analog strongly inhibited their phosphorylation. On the other hand, Ser-43 of Raf-1 was phosphorylated by cGMP in a dose-dependent manner. Therefore, we examined the direct phosphorylation of Raf-1 by PKGII. Wild-type PKGII phosphorylated Raf-1 at Ser-43 in a cGMP-dependent manner, but a PKGII D412A/R415A mutant, which has a low affinity for cGMP, did not. Finally, we found that a phospho-mimic mutant, Raf-1 S43D, suppressed FGF2-induced MAPK pathway. These results suggest that PKGII counters FGF-induced MEK/ERK activation through the phosphorylation of Raf-1 at Ser-43 in chondrocytes.


Subject(s)
Chondrosarcoma/metabolism , Cyclic GMP-Dependent Protein Kinase Type II/metabolism , Fibroblast Growth Factor 2/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Amino Acid Substitution , Animals , Binding Sites , Chondrocytes/metabolism , Cyclic GMP/metabolism , Cyclic GMP-Dependent Protein Kinase Type II/chemistry , Cyclic GMP-Dependent Protein Kinase Type II/genetics , MAP Kinase Signaling System , Mutagenesis, Site-Directed , Phosphorylation , Proto-Oncogene Proteins c-raf/chemistry , Proto-Oncogene Proteins c-raf/genetics , Rats , Serine/chemistry , Signal Transduction , Tumor Cells, Cultured
19.
Int J Hematol ; 105(5): 697-701, 2017 May.
Article in English | MEDLINE | ID: mdl-28000100

ABSTRACT

Abnormal platelet-derived growth factor receptor (PDGFR)-mediated signaling may cause hematologic neoplasm. The PDGFR beta (PDGFRB) gene, located at chromosome band 5q31-33, forms a fusion gene as a result of chromosome translocation. Although patients with PDGFRB rearrangement mostly present with myeloproliferative neoplasm and eosinophilia, acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL) have also been reported in this population. Treatment with imatinib mesylate alone has been shown to have excellent long-term efficacy against myeloproliferative neoplasms; however, its long-term effects on ALL and AML have not been elucidated. A 75-year-old man was diagnosed with acute myeloid leukemia having the PDGFRB and cGMP-dependent protein kinase 2 fusion gene with additional genetic abnormalities. Continuous therapy with single-agent imatinib mesylate resulted in cytogenetic remission and decreased molecular burden for 9 months; however, the leukemia subsequently recurred, and the patient died 1 year after initiation of treatment. This case report supports the importance of cytogenetic analysis during patient screening.


Subject(s)
Antineoplastic Agents/therapeutic use , Imatinib Mesylate/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Receptor, Platelet-Derived Growth Factor beta/genetics , Aged , Chromosomes, Human, Pair 5/genetics , Cyclic GMP-Dependent Protein Kinase Type II/genetics , Cytogenetic Analysis , Fatal Outcome , Gene Fusion/genetics , Gene Rearrangement/genetics , Humans , Male , Receptor, Platelet-Derived Growth Factor beta/physiology , Remission Induction , Signal Transduction/genetics , Translocation, Genetic/genetics , Treatment Outcome
20.
Learn Mem ; 23(8): 435-41, 2016 08.
Article in English | MEDLINE | ID: mdl-27421896

ABSTRACT

Phosphorylation of GluA1, a subunit of AMPA receptors (AMPARs), is critical for AMPAR synaptic trafficking and control of synaptic transmission. cGMP-dependent protein kinase II (cGKII) mediates this phosphorylation, and cGKII knockout (KO) affects GluA1 phosphorylation and alters animal behavior. Notably, GluA1 phosphorylation in the KO hippocampus is increased as a functional compensation for gene deletion, while such compensation is absent in the prefrontal cortex. Thus, there are brain region-specific effects of cGKII KO on AMPAR trafficking, which could affect animal behavior. Here, we show that GluA1 phosphorylation levels differ in various brain regions, and specific behaviors are altered according to region-specific changes in GluA1 phosphorylation. Moreover, we identified distinct regulations of phosphatases in different brain regions, leading to regional heterogeneity of GluA1 phosphorylation in the KO brain. Our work demonstrates region-specific changes in GluA1 phosphorylation in cGKII KO mice and corresponding effects on cognitive performance. We also reveal distinct regulation of phosphatases in different brain region in which region-specific effects of kinase gene KO arise and can selectively alter animal behavior.


Subject(s)
Behavior, Animal/physiology , Brain/metabolism , Cyclic GMP-Dependent Protein Kinase Type II/metabolism , Protein Transport , Receptors, AMPA/metabolism , Amygdala/metabolism , Animals , Conditioning, Classical , Cyclic GMP-Dependent Protein Kinase Type II/genetics , Depression/physiopathology , Fear/physiology , Hippocampus/metabolism , Male , Mice, Inbred C57BL , Olfactory Bulb/metabolism , Phosphorylation , Prefrontal Cortex/metabolism , Smell/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...